Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 3 de 3
Filtrar
1.
Sci Rep ; 12(1): 5938, 2022 04 08.
Artigo em Inglês | MEDLINE | ID: mdl-35396524

RESUMO

Vascular smooth muscle cell (VSMC) proliferation is essential for arteriogenesis to restore blood flow after artery occlusion, but the mechanisms underlying this response remain unclear. Based on our previous findings showing increased VSMC proliferation in the neonatal aorta of mice lacking the protease MT4-MMP, we aimed at discovering new players in this process. We demonstrate that MT4-MMP absence boosted VSMC proliferation in vitro in response to PDGF-BB in a cell-autonomous manner through enhanced p38 MAPK activity. Increased phospho-p38 in basal MT4-MMP-null VSMCs augmented the rate of mitochondrial degradation by promoting mitochondrial morphological changes through the co-activator PGC1α as demonstrated in PGC1α-/- VSMCs. We tested the in vivo implications of this pathway in a novel conditional mouse line for selective MT4-MMP deletion in VSMCs and in mice pre-treated with the p38 MAPK activator anisomycin. Priming of p38 MAPK activity in vivo by the absence of the protease MT4-MMP or by anisomycin treatment led to enhanced arteriogenesis and improved flow recovery after femoral artery occlusion. These findings may open new therapeutic opportunities for peripheral vascular diseases.


Assuntos
Metaloproteinase 17 da Matriz , Proteínas Quinases p38 Ativadas por Mitógeno , Animais , Anisomicina , Proliferação de Células/fisiologia , Células Cultivadas , Metaloproteinase 17 da Matriz/metabolismo , Camundongos , Dinâmica Mitocondrial , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/metabolismo , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/genética , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
2.
Elife ; 92020 10 16.
Artigo em Inglês | MEDLINE | ID: mdl-33063665

RESUMO

Macrophages (Mφs) produce factors that participate in cardiac repair and remodeling after myocardial infarction (MI); however, how these factors crosstalk with other cell types mediating repair is not fully understood. Here we demonstrated that cardiac Mφs increased the expression of Mmp14 (MT1-MMP) 7 days post-MI. We selectively inactivated the Mmp14 gene in Mφs using a genetic strategy (Mmp14f/f:Lyz2-Cre). This conditional KO (MAC-Mmp14 KO) resulted in attenuated post-MI cardiac dysfunction, reduced fibrosis, and preserved cardiac capillary network. Mechanistically, we showed that MT1-MMP activates latent TGFß1 in Mφs, leading to paracrine SMAD2-mediated signaling in endothelial cells (ECs) and endothelial-to-mesenchymal transition (EndMT). Post-MI MAC-Mmp14 KO hearts contained fewer cells undergoing EndMT than their wild-type counterparts, and Mmp14-deficient Mφs showed a reduced ability to induce EndMT in co-cultures with ECs. Our results indicate the contribution of EndMT to cardiac fibrosis and adverse remodeling post-MI and identify Mφ MT1-MMP as a key regulator of this process.


Assuntos
Endotélio Vascular/metabolismo , Transição Epitelial-Mesenquimal , Macrófagos/metabolismo , Metaloproteinase 14 da Matriz/metabolismo , Infarto do Miocárdio/metabolismo , Fator de Crescimento Transformador beta1/metabolismo , Animais , Colágeno/metabolismo , Modelos Animais de Doenças , Feminino , Fibrose , Citometria de Fluxo , Regulação da Expressão Gênica , Células HEK293 , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microcirculação , Fenótipo , Traumatismo por Reperfusão , Disfunção Ventricular Esquerda
3.
Methods Mol Biol ; 1731: 193-212, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29318555

RESUMO

The vasculature ensures optimal delivery of nutrients and oxygen throughout the body. The ability to respond to changing tissue demands requires constant reshaping of the vascular network through modulation of its density, diameter, or patterning. These processes are especially prominent after tissue damage or in tumors. The matrix metalloproteinase (MMP) family of endopeptidases are key contributors to vascular remodeling, able to cleave all extracellular matrix components and also soluble factors and membrane receptors. Observations recorded over several decades have established that the vasculature changes in pathological contexts, and this has formed the basis for developing angiotherapies as a novel approach to treating disease. For example, inhibition of angiogenesis or normalization of the vasculature has been proposed as treatment for cancer and chronic inflammatory diseases. In contrast, boosting angiogenesis may be helpful in ischemic conditions such as myocardial infarction and in regenerative medicine. Classical histological methods for the analysis of tissue vasculature have relied on thin sections that do not capture the complex 3D structure of the vascular network. Given the importance of understanding disease-associated vascular changes for the development of rational angiotherapeutic interventions, we present a protocol for thick section-based 3D image analysis of vasculature structure and function.


Assuntos
Processamento de Imagem Assistida por Computador/métodos , Imageamento Tridimensional/métodos , Metaloproteinase 14 da Matriz/análise , Microvasos/diagnóstico por imagem , Imagem Molecular/métodos , Animais , Linhagem Celular Tumoral , Humanos , Processamento de Imagem Assistida por Computador/instrumentação , Imageamento Tridimensional/instrumentação , Metaloproteinase 14 da Matriz/química , Metaloproteinase 14 da Matriz/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Microscopia Confocal/instrumentação , Microscopia Confocal/métodos , Microtomia , Microvasos/metabolismo , Imagem Molecular/instrumentação , Coloração e Rotulagem/instrumentação , Coloração e Rotulagem/métodos , Ensaios Antitumorais Modelo de Xenoenxerto
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA